<<

RANDOMISED EVALUATION OF COVID-19 THERAPY (RECOVERY)

Background: In early 2020, as this protocol was being developed, there were no approved treatments for COVID-19, a disease induced by the novel coronavirus SARS-CoV-2 that emerged in China in late 2019. The UK New and Emerging Respiratory Virus Threats Advisory Group (NERVTAG) advised that several possible treatments should be evaluated, including -, low-dose corticosteroids, and (which has now been done). A World Health Organization (WHO) expert group issued broadly similar advice. These groups also advised that other treatments will soon emerge that require evaluation.

Eligibility and randomisation: This protocol describes a randomised trial among patients hospitalised for COVID-19. All eligible patients are randomly allocated between several treatment arms, each to be given in addition to the usual standard of care in the participating hospital: No additional treatment vs vs corticosteroids (children only) vs intravenous immunoglobulin (children only). In a factorial design (in the UK alone), eligible patients are allocated simultaneously to no additional treatment vs convalescent plasma vs synthetic neutralising (REGN-COV2). Separately, all participants aged 18 years or older will be allocated to either vs control. The study allows a subsequent randomisation for patients with progressive COVID-19 (evidence of hyper-inflammatory state): No additional treatment vs . For patients for whom not all the trial arms are appropriate or at locations where not all are available, randomisation will be between fewer arms.

Adaptive design: The interim trial results will be monitored by an independent Data Monitoring Committee (DMC). The most important task for the DMC will be to assess whether the randomised comparisons in the study have provided evidence on mortality that is strong enough (with a range of uncertainty around the results that is narrow enough) to affect national and global treatment strategies. In such a circumstance, the DMC will inform the Trial Steering Committee who will make the results available to the public and amend the trial arms accordingly. Regardless, follow-up will continue for all randomised participants, including those previously assigned to trial arms that are modified or ceased. New trial arms can be added as evidence emerges that other candidate therapeutics should be evaluated.

Outcomes: The main outcomes will be death, discharge, need for ventilation and need for renal replacement therapy. For the main analyses, follow-up will be censored at 28 days after randomisation. Additional information on longer term outcomes may be collected through review of medical records or linkage to medical databases where available (such as those managed by NHS Digital and equivalent organisations in the devolved nations).

Simplicity of procedures: To facilitate collaboration, even in hospitals that suddenly become overloaded, patient enrolment (via the internet) and all other trial procedures are greatly streamlined. Informed consent is simple and data entry is minimal. Randomisation via the internet is simple and quick, at the end of which the allocated treatment is displayed on the screen and can be printed or downloaded. Follow-up information is recorded at a single timepoint and may be ascertained by contacting participants in person, by phone or electronically, or by review of medical records and databases.

Page 1 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Data to be recorded: At randomisation, information will be collected on the identity of the randomising clinician and of the patient, age, sex, major co-morbidity, pregnancy, COVID- 19 onset date and severity, and any contraindications to the study treatments. The main outcomes will be death (with date and probable cause), discharge (with date), need for ventilation (with number of days recorded) and need for renal replacement therapy. Reminders will be sent if outcome data have not been recorded by 28 days after randomisation. Suspected Unexpected Serious Adverse Reactions (SUSARs) to one of the study medications (e.g., Stevens-Johnson syndrome, anaphylaxis, aplastic anaemia) will be collected and reported in an expedited fashion. Other adverse events will not be recorded but may be available through linkage to medical databases.

Numbers to be randomised: The larger the number randomised the more accurate the results will be, but the numbers that can be randomised will depend critically on how large the epidemic becomes. If substantial numbers are hospitalised in the participating centres then it may be possible to randomise several thousand with mild disease and a few thousand with severe disease, but realistic, appropriate sample sizes could not be estimated at the start of the trial.

Heterogeneity between populations: If sufficient numbers are studied, it may be possible to generate reliable evidence in certain patient groups (e.g. those with major co-morbidity or who are older). To this end, data from this study may be combined with data from other trials of treatments for COVID-19, such as those being planned by the WHO.

Add-on studies: Particular countries or groups of hospitals, may well want to collaborate in adding further measurements or observations, such as serial virology, serial blood gases or chemistry, serial lung imaging, or serial documentation of other aspects of disease status. While well-organised additional research studies of the natural history of the disease or of the effects of the trial treatments could well be valuable (although the lack of placebo control may bias the assessment of subjective side-effects, such as gastro-intestinal problems), they are not core requirements.

To enquire about the trial, contact the RECOVERY Central Coordinating Office

Nuffield Department of Population Health, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF,

Tel: 0800 1385451 E-mail: [email protected]

Website: www.recoverytrial.net To enquire about the trial outside of the UK, contact the relevant Units (see section 10)

To RANDOMISE a patient, visit:

Website: www.recoverytrial.net

Page 2 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Table of contents 1 BACKGROUND AND RATIONALE ...... 4 1.1 SETTING ...... 4 1.2 TREATMENT OPTIONS ...... 4 1.3 DESIGN CONSIDERATIONS ...... 6 1.4 POTENTIAL FOR EFFECTIVE TREATMENTS TO BECOME AVAILABLE ...... 6 2 DESIGN AND PROCEDURES ...... 7 2.1 ELIGIBILITY...... 7 2.2 CONSENT ...... 8 2.3 BASELINE INFORMATION...... 8 2.4 MAIN RANDOMISATION ...... 9 2.5 SECOND RANDOMISATION FOR PATIENTS WITH PROGRESSIVE COVID-19 ...... 10 2.6 ADMINISTRATION OF ALLOCATED TREATMENT ...... 12 2.7 COLLECTING FOLLOW-UP INFORMATION ...... 12 2.8 DURATION OF FOLLOW-UP ...... 13 2.9 WITHDRAWAL OF CONSENT ...... 14 3 STATISTICAL ANALYSIS ...... 14 3.1 OUTCOMES ...... 14 3.2 METHODS OF ANALYSIS ...... 15 4 DATA AND SAFETY MONITORING ...... 16 4.1 RECORDING SUSPECTED SERIOUS ADVERSE REACTIONS ...... 16 4.2 CENTRAL ASSESSMENT AND ONWARD REPORTING OF SUSARS ...... 16 4.3 RECORDING OTHER ADVERSE EVENTS ...... 17 4.4 ROLE OF THE DATA MONITORING COMMITTEE (DMC) ...... 17 4.5 BLINDING ...... 17 5 QUALITY MANAGEMENT ...... 18 5.1 QUALITY BY DESIGN PRINCIPLES ...... 18 5.2 TRAINING AND MONITORING ...... 18 5.3 DATA MANAGEMENT ...... 19 5.4 SOURCE DOCUMENTS AND ARCHIVING ...... 19 6 OPERATIONAL AND ADMINISTRATIVE DETAILS ...... 19 6.1 SPONSOR AND COORDINATION ...... 19 6.2 FUNDING ...... 20 6.3 INDEMNITY ...... 20 6.4 LOCAL CLINICAL CENTRES ...... 20 6.5 SUPPLY OF STUDY TREATMENTS ...... 20 6.6 END OF TRIAL ...... 21 6.7 PUBLICATIONS AND REPORTS ...... 21 6.8 SUBSTUDIES ...... 21 7 VERSION HISTORY ...... 22 8 APPENDICES ...... 23 8.1 APPENDIX 1: INFORMATION ABOUT THE TREATMENT ARMS ...... 23 8.2 APPENDIX 2: DRUG SPECIFIC CONTRAINDICATIONS AND CAUTIONS ...... 26 8.3 APPENDIX 3: PAEDIATRIC DOSING INFORMATION ...... 29 8.4 APPENDIX 4: USE OF IMPS IN PREGNANT AND BREASTFEEDING WOMEN ...... 31 8.5 APPENDIX 5: ORGANISATIONAL STRUCTURE AND RESPONSIBILITIES ...... 33 8.6 APPENDIX 5: ORGANISATIONAL DETAILS ...... 35 9 REFERENCES ...... 36 10 CONTACT DETAILS ...... 39

Page 3 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

1 BACKGROUND AND RATIONALE

1.1 Setting In 2019 a novel coronavirus-disease (COVID-19) emerged in Wuhan, China. A month later the Chinese Center for Disease Control and Prevention identified a new beta-coronavirus (SARS coronavirus 2, or SARS-CoV-2) as the aetiological agent.1 The clinical manifestations of COVID-19 range from asymptomatic infection or mild, transient symptoms to severe viral pneumonia with respiratory failure. As many patients do not progress to severe disease the overall case fatality rate per infected individual is low, but hospitals in areas with significant community transmission have experienced a major increase in the number of hospitalised pneumonia patients, and the frequency of severe disease in hospitalised patients can be as high as 30%.2-4 The progression from prodrome (usually fever, fatigue and cough) to severe pneumonia requiring oxygen support or often takes one to two weeks after the onset of symptoms.2 The kinetics of viral replication in the respiratory tract are not well characterized, but this relatively slow progression provides a potential time window in which antiviral therapies could influence the course of disease. In May 2020 a new COVID-associated inflammatory syndrome in children was identified, Paediatric Inflammatory Multisystem Syndrome - Temporally associated with SARS-CoV-2 (PIMS-TS).5 A rapid NHS England-led consensus process identified the need to evaluate corticosteroids and intravenous immunoglobulin (IVIg) as initial therapies in PIMS-TS, and confirmed tocilizumab as one of the biological anti-inflammatory agents to be evaluated as a second line therapy.

1.2 Treatment Options

1.2.1 Main randomisation This protocol allows reliable assessment of the effects of multiple different treatments (including re-purposed and novel drugs) on major outcomes in COVID-19. All patients will receive usual care for the participating hospital.

Randomisation part A: Eligible patients may be randomly allocated between the following treatment arms:

 No additional treatment

 Colchicine (men ≥18 years old and women ≥55 years old only)

 Corticosteroids (children ≤44 weeks gestational age, or >44 weeks gestational age with PIMS-TS only)

 Intravenous immunoglobulin (children >44 weeks gestational age with PIMS- TS only)

Page 4 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Randomisation part B [UK only]: Simultaneously, eligible patients will be randomly allocated between the following treatment arms:

 No additional treatment

 Convalescent plasma

 Synthetic neutralising antibodies (REGN-COV2) (adults and children ≥12 years old only)

Randomisation part C (adults ≥18 years old only): Simultaneously, eligible patients will be randomly allocated between the following treatment arms:

 No additional treatment

 Aspirin

1.2.2 Second randomisation for patients with progressive COVID-19 Severe COVID-19 is associated with release of pro-inflammatory cytokines, such as IL-1, IL-6 and TNFα, and other markers of systemic including ferritin and C-reactive protein.6-8 There is a possibility that this response may cause or exacerbate lung injury, leading to life-threatening disease.

Participants with progressive COVID-19 (as evidenced by hypoxia and an inflammatory state) may undergo an optional second randomisation between the following treatment arms:

 No additional treatment

 Tocilizumab

1.2.3 Modifications to the number of treatment arms Other arms can be added to the first or second randomisation if evidence emerges that there are suitable candidate therapeutics. Conversely, in some patient populations, not all trial arms are appropriate (e.g. due to contraindications based on co-morbid conditions or concomitant medication); in some hospitals or countries, not all treatment arms will be available (e.g. due to manufacturing and supply shortages); and at some times, not all treatment arms will be active (e.g. due to lack of relevant approvals and contractual agreements). The Trial Steering Committee may elect to pause one or more of the arms in order to increase trial efficiency during a fluctuating epidemic. In any of these situations, randomisation will be between fewer arms. Depending on the availability and suitability of treatments, it may be allowed for participants to be randomised in only one or two parts (A, B, or C) of the main randomisation.

Page 5 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

1.3 Design Considerations The RECOVERY Protocol describes an overarching trial design to provide reliable evidence on the efficacy of candidate therapies for suspected or confirmed COVID-19 infection in hospitalised patients receiving usual standard of care.

In early 2020, when the trial first started, there were no known treatments for COVID-19. The anticipated scale of the epidemic is such that hospitals, and particularly intensive care facilities, may be massively overstretched at some points in time, with around 10% requiring hospitalisation. In this situation, even treatments with only a moderate impact on survival or on hospital resources could be worthwhile. Therefore, the focus of RECOVERY is the impact of candidate treatments on mortality and on the need for hospitalisation or ventilation.

Critically, the trial is designed to minimise the burden on front-line hospital staff working within an overstretched care system during a major epidemic. Eligibility criteria are therefore simple and trial processes (including paperwork) are minimised.

The protocol is deliberately flexible so that it is suitable for a wide range of settings, allowing:  a broad range of patients to be enrolled in large numbers;  randomisation between only those treatment arms that are both available at the hospital and not believed by the enrolling doctor to be contraindicated (e.g. by particular co-morbid conditions or concomitant medications);  treatment arms to be added or removed according to the emerging evidence; and  additional substudies may be added to provide more detailed information on side effects or sub-categorisation of patient types but these are not the primary objective and are not required for participation.

In a cohort of 191 hospitalised COVID-19 patients with a completed outcome, the median time from illness onset to discharge was 22·0 days (IQR 18·0–25·0) and the median time to death was 18·5 days (15·0–22·0). Thirty-two patients (17%) required invasive mechanical ventilation and the median time from onset to mechanical ventilation was 14.5 days. Therefore, early endpoint assessment, such as 28 days after randomisation, is likely to provide largely complete outcome data and will permit early assessment of treatment efficacy and safety.9

1.4 Potential for effective treatments to become available

In early 2020, when the trial first started, there were no known treatments for COVID-19. However, over time, effective treatments may become available, typically as the result of reliable information from randomised trials (including from this study). For example, in June 2020, results from the RECOVERY trial showed that reduces the mortality in COVID-19 patients requiring mechanical ventilation or oxygen. In response, many clinical guidelines now recommend the use of dexamethasone as standard of care for these types of patients.

The RECOVERY trial randomises eligible participant to usual standard of care for the local hospital alone vs usual standard of care plus one or more additional study treatments. Over time, it is expected that usual standard of care alone will evolve. Thus randomisation will always be relevant to the current clinical situation and the incremental effects of the study treatments will be appropriately assessed. Page 6 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

2 DESIGN AND PROCEDURES

2.1 Eligibility Patients are eligible for the study if all of the following are true:

(i) Hospitalised

(ii) SARS-CoV-2 infection associated disease (clinically suspected or laboratory confirmed)

In general, SARS-CoV-2 infection should be suspected when a patient presents with: a) typical symptoms (e.g. influenza-like illness with fever and muscle pain, or respiratory illness with cough and shortness of breath); and b) compatible chest X-ray findings (consolidation or ground-glass shadowing); and c) alternative causes have been considered unlikely or excluded (e.g. heart failure, influenza). However, the diagnosis remains a clinical one based on the opinion of the managing doctor.

A small number of children (aged <18 years) present with atypical features, including a hyperinflammatory state and evidence of single or multi-organ dysfunction (called Paediatric Multisystem Inflammatory Syndrome temporally associated with COVID- 19 [PIMS-TS]). Some do not have significant lung involvement.a

(iii) No medical history that might, in the opinion of the attending clinician, put the patient at significant risk if he/she were to participate in the trial

In addition, if the attending clinician believes that there is a specific contra-indication to one of the active drug treatment arms (see Appendix 2; section 8.2 and Appendix 3; section 8.3 for children) or that the patient should definitely be receiving one of the active drug treatment arms then that arm will not be available for randomisation for that patient. For patients who lack capacity, an advanced directive or behaviour that clearly indicates that they would not wish to participate in the trial would be considered sufficient reason to exclude them from the trial.

In some locations, children (aged <18 years) will not be recruited, to comply with local and national regulatory approvals (see Section 8.3).

a https://www.rcpch.ac.uk/sites/default/files/2020-05/COVID-19-Paediatric-multisystem-%20inflammatory%20syndrome- 20200501.pdf Page 7 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

2.2 Consent Informed consent should be obtained from each patient 16 years and over before enrolment into the study. However, if the patient lacks capacity to give consent due to the severity of their medical condition (e.g. acute respiratory failure or need for immediate ventilation) or prior disease, then consent may be obtained from a relative acting as the patient’s legally designated representative or – if a suitable relative is not available after reasonable efforts to locate one – an independent doctor. Further consent will then be sought with the patient if they recover sufficiently. For children aged <16 years old consent will be sought from their parents or legal guardian. Where possible, children aged between 10-15 years old will also be asked for assent. Children aged ≥16 years old will asked for consent as for adults. Witnessed consent may be obtained over the telephone or web video link if hospital visiting rules or parental infection mean a parent/guardian cannot be physically present.

Due to the poor outcomes in COVID-19 patients who require ventilation (>90% mortality in one cohort9), patients who lack capacity to consent due to severe disease (e.g. needs ventilation), and for whom a relative to act as the legally designated representative is not available, randomisation and consequent treatment will proceed with consent provided by a treating clinician (independent of the clinician seeking to enrol the patient) who will act as the legally designated representative (if allowed by local regulations). Consent will then be obtained from the patient’s personal legally designated representative (or directly from the patient if they recover promptly) at the earliest opportunity.

2.3 Baseline information The following information will be recorded on the web-based form by the attending clinician or delegate:

 Patient details (e.g. name or initials [depending on privacy requirements], NHS/CHI number [UK only] or medical records number, date of birth, sex)  Clinician details (e.g. name)  COVID-19 symptom onset date  COVID-19 severity as assessed by need for supplemental oxygen, non-invasive ventilation or invasive mechanical ventilation/extracorporeal membrane oxygenation (ECMO)  Oxygen saturations on air (if available)  Latest routine measurement of creatinine, C-reactive protein, and D-dimer (if available)  SARS-CoV-2 PCR test result (if available)  Major co-morbidity (e.g. heart disease, diabetes, chronic lung disease) and pregnancy  Use of relevant medications (corticosteroids, remdesivir, antiplatelet and anticoagulant therapy)  Date of hospitalisation  Contraindication to the study treatment regimens (in the opinion of the attending clinician)  Willingness to receive a blood product [UK only]  Name of person completing the form

The person completing the form will then be asked to confirm that they wish to randomise the patient and will then be required to enter their name and e-mail address.

Page 8 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

2.4 Main randomisation In addition to receiving usual care, eligible patients will be allocated using a central web- based randomisation service (without stratification or minimisation). From version 6.0 of the protocol, a factorial design will be used such that eligible patients may be randomised to one of the treatment arms in Randomisation A and, simultaneously, to one of the treatment arms in Randomisation B. From version 10.0 of the protocol, a further factorial randomisation was added (Main Randomisation part C). From version 11.3 of the protocol, children may be recruited into the trial even if there are no main randomisation treatments which are both available and suitable provided they meet the criteria for inclusion in the second randomisation, per section 2.5. They will not be allocated to a main randomisation group, but will be potentially eligible for the second randomisation between tocilizumab and control.

2.4.1 Main randomisation part A: Eligible patients may be randomised to one of the arms listed below. The doses in this section are for adults. Please see Appendix 3 for paediatric dosing. Study treatments do not need to be continued after discharge from hospital.

 No additional treatment

 Colchine 1 mg after randomisation followed by 500mcg 12 hours later and then 500 mcg twice daily by mouth or nasogastric tube for 10 days in total.b (Men ≥18 years old and women ≥55 years old only.)

 Corticosteroid (in children ≤44 weeks gestational age, or >44 weeks gestational age with PIMS-TS only): see Appendix 3.

 Intravenous immunoglobulin (in children >44 weeks gestational age with PIMS-TS only): see Appendices 2 and 3 for dose, contraindications and monitoring information.

For randomisation part A, the randomisation program will allocate patients in a ratio of 1:1 between the no additional treatment arm and each of the other arms available. If one or more of the active drug treatments is not available at the hospital or is believed, by the attending clinician, to be contraindicated (or definitely indicated) for the specific patient, then this fact will be recorded via the web-based form prior to randomisation; random allocation will then be between the remaining arms. If no treatments are both available and suitable, then it may be possible to only be randomised in part B (UK only) and/or part C.

2.4.2 Main randomisation part B [UK only]: Eligible patients may be randomised to one of the arms listed below. The doses in this section are for adults. Please see Appendix 3 for paediatric dosing. Participants in this randomisation should have a serum sample sent to their transfusion laboratory prior to randomisation in which presence of antibodies against SARS-CoV-2 may be tested.

 No additional treatment

b Treatment should be discontinued at 10 days or on discharge from hospital if sooner Page 9 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

 Convalescent plasma Single unit of ABO compatible convalescent plasma (275mls +/- 75 mls) intravenous per day on study days 1 (as soon as possible after randomisation) and 2 (with a minimum of 12 hour interval between 1st and 2nd units). ABO identical plasma is preferred if available. The second transfusion should not be given if patient has a suspected serious adverse reaction during or after the first transfusion.

 Synthetic neutralising antibodies (REGN-COV2; adults and children aged ≥12 yearsc only). A single dose of REGN10933 + REGN10987 8 g (4 g of each monoclonal ) in 250ml 0.9% saline infused intravenously over 60 minutes +/- 15 minutes as soon as possible after randomisation

For randomisation part B, the randomisation program will allocate patients in a ratio of 1:1:1 between each of the arms. If the active treatment is not available at the hospital, the patient does not consent to receive convalescent plasma, or is believed, by the attending clinician, to be contraindicated for the specific patient, then this fact will be recorded via the web- based form and the patient will be excluded from the relevant arm in Randomisation part B.

2.4.3 Main randomisation part C [adults aged ≥18 years only]: Eligible patients may be randomised to one of the arms listed below.

 No additional treatment

 Aspirin 150 mg by mouth (or nasogastric tube) or per rectum once daily until discharge.d

Note: The allocation in this randomisation should not influence the use of standard thromboprophylaxis care.

The randomisation program will allocate patients in a ratio of 1:1 between the arms being evaluated in part C of the main randomisation.

2.5 Second randomisation for patients with progressive COVID-19 Patients enrolled in the RECOVERY trial and with clinical evidence of a hyper-inflammatory state may be considered for a second randomisation if they meet the following criteria:

(i) Recruited into the RECOVERY trial no more than 21 days agoe (ii) Clinical evidence of progressive COVID-19: a. oxygen saturation <92% on room air or requiring oxygen (or in children (age <18 years), significant systemic disease with persistent pyrexia, with or without evidence of respiratory involvement)f; and

c Older children who weigh <40kg will also not be eligible for this treatment. d In countries where 150mg aspirin is not available, up to 162mg may be used instead e Children recruited into RECOVERY for whom no main randomisation treatment are both available and suitable (see section 2.4) should undergo this second randomisation as soon as possible after recruitment. f A small number of children (age <18 years) present with atypical features, including a hyperinflammatory state and evidence of single or multi-organ dysfunction. Some do not have significant lung involvement.

Page 10 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

b. C-reactive protein ≥75 mg/L (iii) No medical history that might, in the opinion of the attending clinician, put the patient at significant risk if he/she were to participate in this aspect of the RECOVERY trial. (Note: Pregnancy and breastfeeding are not specific exclusion criteria.)

Note: Participants may undergo this second randomisation at any point after being first randomised, provided they meet the above criteria, and thus may receive up to four study treatments (one each from Main randomisation parts A, B and C, plus one from the second randomisation). For some participants the second randomisation may be immediately after the first but for others it may occur a few hours or days later, if and when they deteriorate.

The following information will be recorded (on the web-based form) by the attending clinician or delegate:

 Patient details (e.g. name or initials, NHS/CHI number [UK only] or medical records number, date of birth, sex)  Clinician details (e.g. name)  COVID-19 severity as assessed by need for supplemental oxygen or ventilation/ECMO  Markers of progressive COVID-19 (including oxygen saturation, C-reactive protein)  Contraindication to the study drug treatments (in the opinion of the attending clinician)  Name of person completing the form

The person completing the form will then be asked to confirm that they wish to randomise the patient and will then be required to enter their own name and e-mail address.

Eligible participants may be randomised between the following treatment arms:

 No additional treatment

 Tocilizumab by intravenous infusion with the dose determined by body weight:

Weight* Dose >40 and ≤65 kg 400 mg >65 and ≤90 kg 600 mg >90 kg 800 mg * for lower weights, dosing should be 8 mg/kg (see Appendix 3 for paediatric dosing) (Note: body weight may be estimated if it is impractical to weigh the patient)

Tocilizumab should be given as a single intravenous infusion over 60 minutes in 100ml sodium chloride 0.9%. A second dose may be given ≥12 and <24 hours later if, in the opinion of the attending clinician, the patient’s condition has not improved.

The randomisation program will allocate patients in a ratio of 1:1 between the arms being evaluated in the second randomisation. Participants should receive standard management

(see: https://www.rcpch.ac.uk/sites/default/files/2020-05/COVID-19-Paediatric-multisystem- %20inflammatory%20syndrome-20200501.pdf) Page 11 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

(including blood tests such as liver function tests and full blood count) according to their clinical need.

2.6 Administration of allocated treatment The details of the allocated study treatments will be displayed on the screen and can be printed or downloaded. The hospital clinicians are responsible for prescription and administration of the allocated treatments. The patient’s own doctors are free to modify or stop study treatments if they feel it is in the best interests of the patient without the need for the patient to withdraw from the study (see section 2.9). This study is being conducted within hospitals. Therefore use of medication will be subject to standard medication reviews (typically within 48 hours of enrolment) which will guide modifications to both the study treatment and use of concomitant medication (e.g. in the case of potential drug interactions).

Note: [UK only] NHS guidelines require patients to have two separate blood samples taken for Group and Screen prior to administration of blood products. Each sample is approximately 5 mL and both need to be taken at any time between admission to hospital and receipt of the first plasma transfusion (as the laboratory will not issue plasma without both samples), although if a valid historical sample exists this can be used for one of the samples. The participant’s blood group is identified to ensure that blood group-compatible plasma is given and this information would be available to the participant if they wish. Such tests may be required as part of the routine care of the participant if the managing team wish to consider using blood products and samples will be stored, retained and destroyed as per trust standard procedures and protocols. The extra serum sample collected for measurement of coronavirus and antibodies against it will be prepared in the local transfusion laboratory (including removing any identifiers and labelling with the participant’s study ID) and sent to a central laboratory for analysis. Once testing is complete these samples will be destroyed.

2.7 Collecting follow-up information The following information will be ascertained at the time of death or discharge or at 28 days after first randomisation (whichever is sooner):  Vital status (alive / dead, with date and presumed cause of death, if appropriate)  Hospitalisation status (inpatient / discharged, with date of discharge, if appropriate)  SARS-CoV-2 test result  Use of ventilation (with days of use and type, if appropriate)  Use of renal dialysis or haemofiltration  Documented new major cardiac arrhythmia (including atrial and ventricular arrhythmias)  Major bleeding (defined as intracranial bleeding or bleeding requiring transfusion, endoscopy, surgery, or vasoactive drugs)  Thrombotic event, defined as either (i) acute pulmonary embolism; (ii) deep vein thrombosis; (iii) ischaemic stroke; (iv) myocardial infarction; or (v) systemic arterial embolism.  Use of any medications included in the RECOVERY trial protocol (including drugs in the same class) or other purported COVID-19 treatments (e.g. remdesivir, favipiravir)  Participation in other randomised trials of interventions (vaccines or treatments) for COVID-19.

Page 12 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

 Additional information including results of routine tests (including full blood count, coagulation and inflammatory markers, cardiac biomarkers, electro- and echo- cardiograms) and other treatments given will be collected for children in the UK. This information will be obtained and entered into the web-based IT system by a member of the hospital clinical or research staff. At some locations, electrocardiograms done as part of routine care of adult participants will also be collected.

Follow-up information is to be collected on all study participants, irrespective of whether or not they complete the scheduled course of allocated study treatment. Study staff will seek follow-up information through various means including medical staff, reviewing information from medical notes, routine healthcare systems, and registries.

For all randomised participants, vital status (alive / dead, with date and presumed cause of death, if appropriate) is to be ascertained at 28 days after first randomisation. This may be achieved through linkage to routine death registration data (e.g. in the UK) or through direct contact with the participant, their relatives, or medical staff and completion of an additional follow-up form.

2.7.1 Additional assessment of safety of antibody-based therapy [UK only] For at least the first 200 participants in each comparison in Main Randomisation part B (no additional treatment vs. convalescent plasma and no additional treatment vs. synthetic neutralising antibody), the following information will be collected on the following events occurring within the first 72 hours after randomisation:  Sudden worsening in respiratory status  Severe allergic reaction or other infusion reaction  Temperature >39oC or ≥2oC rise above baseline  Sudden hypotension, defined as either (i) sudden drop in systolic blood pressure of ≥30 mmHg with systolic blood pressure ≤80 mmHg; or (ii) requiring urgent medical attention  Clinical haemolysis, defined as fall in haemoglobin plus one or more of the following: rise in lactate dehydrogenase (LDH), rise in bilirubin, positive direct antiglobulin test (DAT), or positive crossmatch  Thrombotic event, defined as either (i) acute pulmonary embolism; or (ii) deep-vein thrombosis; or (iii) ischaemic stroke; or (iv) myocardial infarction; or (v) systemic arterial embolism.

The Data Monitoring Committee will review unblinded information on these outcomes and advise if, in their view, the collection of such information should be extended to more participants.

In addition, Serious Hazards Of Transfusion (SHOT) reporting will be conducted for all patients receiving convalescent plasma for the full duration of the study (see section 4.1).

2.8 Duration of follow-up All randomised participants are to be followed up until death, discharge from hospital or 28 days after randomisation (whichever is sooner). It is recognised that in the setting of this trial, there may be some variability in exactly how many days after randomisation, Page 13 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

information on disease status is collected. This is acceptable and will be taken account of in the analyses and interpretation of results, the principle being that some information about post-randomisation disease status is better than none.

In the UK, longer term (up to 10 years) follow-up will be sought through linkage to electronic healthcare records and medical databases including those held by NHS Digital, Public Health England and equivalent bodies, and to relevant research databases (e.g. UK Biobank, Genomics England).

2.9 Withdrawal of consent A decision by a participant (or their parent/guardian) that they no longer wish to continue receiving study treatment should not be considered to be a withdrawal of consent for follow- up. However, participants (or their parent/guardian) are free to withdraw consent for some or all aspects of the study at any time if they wish to do so. In accordance with regulatory guidance, de-identified data that have already been collected and incorporated in the study database will continue to be used (and any identifiable data will be destroyed).

For participants who lack capacity, if their legal representative withdraws consent for treatment or methods of follow-up then these activities would cease.

3 STATISTICAL ANALYSIS

All analyses for reports, presentations and publications will be prepared by the coordinating centre at the Nuffield Department of Population Health, University of Oxford. A more detailed statistical analysis plan will be developed by the investigators and published on the study website whilst still blind to any analyses of aggregated data on study outcomes by treatment allocation.

3.1 Outcomes For each pairwise comparison with the ‘no additional treatment’ arm, the primary objective is to provide reliable estimates of the effect of study treatments on all-cause mortality at 28 days after randomisation (with subsidiary analyses of cause of death and of death at various timepoints following discharge).

The secondary objectives are to assess the effects of study treatments on duration of hospital stay; and, among patients not on invasive mechanical ventilation at baseline, the composite endpoint of death or need for invasive mechanical ventilation or ECMO.

Other objectives include the assessment of the effects of study treatments on the need for any ventilation (and duration of invasive mechanical ventilation), renal replacement therapy and thrombotic events. Safety outcomes include bleeding, new major cardiac arrhythmias and (assessed at 72 hours after randomization among participants in main randomization part B only) sudden worsening in respiratory status, severe allergic reaction, significant fever, sudden hypotension and clinical haemolysis.

Study outcomes will be assessed based on data recorded up to 28 days and up to 6 months after randomisation.

Page 14 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Where available, data from routine healthcare records (including linkage to medical databases held by organisations such as NHS Digital in the UK) and from relevant research studies (such as UK Biobank, Genomics England, ISARIC-4C and PHOSP-COVID) will allow subsidiary analyses of the effect of the study treatments on particular non-fatal events (e.g. ascertained through linkage to Hospital Episode Statistics), the influence of pre-existing major co-morbidity (e.g. diabetes, heart disease, lung disease, hepatic insufficiency, severe depression, severe kidney impairment, immunosuppression), and longer-term outcomes as well as in particular sub-categories of patient (e.g. by genotype, pregnancy).

3.2 Methods of analysis For all outcomes, comparisons will be made between all participants randomised to the different treatment arms, irrespective of whether they received their allocated treatment (“intention-to-treat” analyses).

For time-to-event analyses, each treatment group will be compared with the no additional treatment group using the log-rank test. Kaplan-Meier estimates for the time to event will also be plotted (with associated log-rank p-values). The log-rank ‘observed minus expected’ statistic (and its variance) will also be used to estimate the average event rate ratio (and its confidence interval) for those allocated to each treatment group versus the no additional treatment group. For binary outcomes where the timing is unknown, the risk ratio and absolute risk difference will be calculated with confidence intervals and p-value reported. For the primary outcome (death within 28 days of randomisation), discharge alive before 28 days will assume safety from the event (unless there is additional data confirming otherwise).

Pairwise comparisons within each randomisation will be made between each treatment arm and the no additional treatment arm (reference group) in that particular randomisation (main randomisation part A, B or C, and second randomisation). However, since not all treatments may be available or suitable for all patients, those in the no additional treatment arm will only be included in a given comparison if, at the point of their randomisation, they could alternatively have been randomised to the active treatment of interest. Allowance for multiple treatment comparisons due to the multi-arm design will be made. All p-values will be 2-sided.

Pre-specified subgroup analysis (e.g., level of respiratory support, time since onset of symptoms; sex; age group; ethnicity; use of corticosteroids) will be conducted for the primary outcome using the statistical test for interaction (or test for trend where appropriate). Sensitivity analyses will be conducted among those patients with laboratory confirmed SARS-CoV-2.

Further details will be fully described in the Statistical Analysis Plan.

Page 15 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

4 DATA AND SAFETY MONITORING

4.1 Recording Suspected Serious Adverse Reactions The focus is on those events that, based on a single case, are highly likely to be related to the study medication. Examples include anaphylaxis, Stevens Johnson Syndrome, or bone marrow failure, where there is no other plausible explanation.

Any Serious Adverse Eventg that is believed with a reasonable probability to be due to one of the study treatments will be considered a Suspected Serious Adverse Reaction (SSAR). In making this assessment, there should be consideration of the probability of an alternative cause (for example, COVID-19 itself or some other condition preceding randomisation), the timing of the event with respect to study treatment, the response to withdrawal of the study treatment, and (where appropriate) the response to subsequent re-challenge.

All SSARs should be reported by telephone to the Central Coordinating Office and recorded on the study IT system immediately.

[UK only] Suspected serious transfusion reactions in patients who receive convalescent plasma should additionally be reported to Serious Hazards of Transfusions (SHOT) and through the MHRA Serious Adverse Blood Reactions and Events (SABRE) system.h

4.2 Central assessment and onward reporting of SUSARs

Clinicians at the Central Coordinating Office are responsible for expedited review of reports of SSARs received. Additional information (including the reason for considering it both serious and related, and relevant medical and medication history) will be sought.

The focus of Suspected Unexpected Serious Adverse Reaction (SUSAR) reporting will be on those events that, based on a single case, are highly likely to be related to the study medication. To this end, anticipated events that are either efficacy endpoints, consequences of the underlying disease, or common in the study population will be exempted from expedited reporting. Thus the following events will be exempted from expedited reporting:

(i) Events which are the consequence of COVID-19; and (ii) Common events which are the consequence of conditions preceding randomisation.

Any SSARs that are not exempt will be reviewed by a Central Coordinating Office clinician and an assessment made of whether the event is “expected” or not (assessed against the relevant Summary of Product Characteristics or Investigator Brochure). Any SSARs that are not expected would be considered a Suspected Unexpected Serious Adverse Reaction (SUSAR).

g Serious Adverse Events are defined as those adverse events that result in death; are life-threatening; require in-patient hospitalisation or prolongation of existing hospitalisation; result in persistent or significant disability or incapacity; result in congenital anomaly or birth defect; or are important medical events in the opinion of the responsible investigator (that is, not life-threatening or resulting in hospitalisation, but may jeopardise the participant or require intervention to prevent one or other of the outcomes listed above). h https://www.shotuk.org/reporting/ Page 16 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

All confirmed SUSARs will be reported to the Chair of the DMC and to relevant regulatory authorities, ethics committees, and investigators in an expedited manner in accordance with regulatory requirements.

4.3 Recording other Adverse Events In addition to recording Suspected Serious Adverse Reactions (see section 4.1), information will be collected on all deaths and efforts will be made to ascertain the underlying cause. Other serious or non-serious adverse events will not be recorded unless specified in section 2.7. It is anticipated that for some substudies, more detailed information on adverse events (e.g. through linkage to medical databases) or on other effects of the treatment (e.g. laboratory or radiological features) will be recorded and analysed but this is not a requirement of the core protocol.

4.4 Role of the Data Monitoring Committee (DMC) During the study, interim analyses of all study data will be supplied in strict confidence to the independent DMC. The DMC will request such analyses at a frequency relevant to the emerging data from this and other studies.

The DMC will independently evaluate these analyses and any other information considered relevant. The DMC will determine if, in their view, the randomised comparisons in the study have provided evidence on mortality that is strong enough (with a range of uncertainty around the results that is narrow enough) to affect national and global treatment strategies. In such a circumstance, the DMC will inform the Trial Steering Committee who will make the results available to the public and amend the trial arms accordingly. Unless this happens, the Trial Steering Committee, Chief Investigator, study staff, investigators, study participants, funders and other partners will remain blind to the interim results until 28 days after the last patient has been randomised for a particular intervention arm (at which point analyses may be conducted comparing that arm with the no additional treatment arm).

The DMC will review the safety and efficacy analyses among children (age <18 years) both separately and combined with the adult data. As described in section 2.7.1, the DMC will advise if collection of information relating to the safety of convalescent plasma should be extended beyond the first 200 patients enrolled to each comparison in Main Randomisation part B.

4.5 Blinding This is an open-label study. However, while the study is in progress, access to tabular results of study outcomes by allocated treatment allocation will not be available to the research team, patients, or members of the Trial Steering Committee (unless the DMC advises otherwise).

Page 17 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

5 QUALITY MANAGEMENT

5.1 Quality By Design Principles In accordance with the principles of Good Clinical Practice and the recommendations and guidelines issued by regulatory agencies, the design, conduct and analysis of this trial is focussed on issues that might have a material impact on the wellbeing and safety of study participants (hospitalised patients with suspected or confirmed SARS-CoV-2 infection) and the reliability of the results that would inform the care for future patients.

The critical factors that influence the ability to deliver these quality objectives are:  to minimise the burden on busy clinicians working in an overstretched hospital during a major epidemic  to ensure that suitable patients have access to the trial medication without impacting or delaying other aspects of their emergency care  to provide information on the study to patients and clinicians in a timely and readily digestible fashion but without impacting adversely on other aspects of the trial or the patient’s care  to allow individual clinicians to use their judgement about whether any of the treatment arms are not suitable for the patient  to collect comprehensive information on the mortality and disease status

In assessing any risks to patient safety and well-being, a key principle is that of proportionality. Risks associated with participation in the trial must be considered in the context of usual care. At present, there are no proven treatments for COVID-19, basic hospital care (staffing, beds, ventilatory support) may well be overstretched, and mortality for hospitalised patients may be around 10% (or more in those who are older or have significant co-morbidity).

5.2 Training and monitoring The focus will be on those factors that are critical to quality (i.e. the safety of the participants and the reliability of the trial results). Remedial actions would focus on issues with the potential to have a substantial impact on the safety of the study participants or the reliability of the results.

The study will be conducted in accordance with the principles of International Conference on Harmonisation Guidelines for Good Clinical Research Practice (ICH-GCP) and relevant local, national and international regulations. Any serious breach of GCP in the conduct of the clinical trial will be handled in accordance with regulatory requirements. Prior to initiation of the study at each Local Clinical Centre (LCC), the Central Coordinating Office (CCO) or relevant Regional Coordinating Centre (RCC) will confirm that the LCC has adequate facilities and resources to carry out the study. LCC lead investigators and study staff will be provided with training materials.

In the context of this epidemic, visits to hospital sites is generally not appropriate as they could increase the risks of spreading infection, and in the context of this trial they generally would not influence the reliability of the trial results or the well-being of the participants. In exceptional circumstances, the CCO or RCC may arrange monitoring visits to LCCs as considered appropriate based on perceived training needs and the results of central Page 18 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

statistical monitoring of study data.10,11 The purpose of such visits will be to ensure that the study is being conducted in accordance with the protocol, to help LCC staff to resolve any local problems, and to provide extra training focussed on specific needs. No routine source data verification will take place.

In the UK, training of laboratory and transfusion staff and initiation of convalescent plasma delivery will be performed by NHS Blood and Transplant Clinical Trials Unit.

5.3 Data management LCC clinic staff will use the bespoke study web-based applications for study management and to record participant data (including case report forms) in accordance with the protocol. Data will be held in central databases located at the CCO or on secure cloud servers. In some circumstances (e.g. where there is difficulty accessing the internet or necessary IT equipment), paper case report forms may be required with subsequent data entry by either LCC or CCO staff. Although data entry should be mindful of the desire to maintain integrity and audit trails, in the circumstances of this epidemic, the priority is on the timely entry of data that is sufficient to support reliable analysis and interpretation about treatment effects. CCO staff will be responsible for provision of the relevant web-based applications and for generation of data extracts for analyses.

All data access will be controlled by unique usernames and passwords, and any changes to data will require the user to enter their username and password as an electronic signature in accordance with regulatory requirements.12 Staff will have access restricted to the functionality and data that are appropriate for their role in the study.

5.4 Source documents and archiving Source documents for the study constitute the records held in the study main database. These will be retained for at least 25 years from the completion of the study. Identifiable data will be retained only for so long as it is required to maintain linkage with routine data sources (see section 2.8), with the exception of children for whom such data must be stored until they reach 21 years old (due to the statute of limitations). The sponsor and regulatory agencies will have the right to conduct confidential audits of such records in the CCO and LCCs (but should mindful of the workload facing participating hospitals and the infection control requirements during this epidemic).

6 OPERATIONAL AND ADMINISTRATIVE DETAILS

6.1 Sponsor and coordination The University of Oxford will act as the trial Sponsor. The trial will be coordinated by a Central Coordinating Office (CCO) within the Nuffield Department of Population Health staffed by members of the two registered clinical trials units – the Clinical Trial Service Unit and the National Perinatal Epidemiology Unit Clinical Trials Unit. The CCO will oversee Regional Coordinating Centres which will assist with selection of Local Clinical Centres (LCCs) within their region and for the administrative support and monitoring of those LCCs. The data will be collected, analysed and published independently of the source of funding.

Page 19 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

6.2 Funding This study is supported by a grant to the University of Oxford from UK Research and Innovation/National Institute for Health Research (NIHR) and by core funding provided by NIHR Oxford Biomedical Research Centre, the Wellcome Trust, the Bill and Melinda Gates Foundation, Department for International Development, Health Data Research UK, NIHR Health Protection Unit in Emerging and Zoonotic Infections and the Medical Research Council Population Health Research Unit, and NIHR Clinical Trials Unit Support Funding.

6.3 Indemnity The University has a specialist insurance policy in place which would operate in the event of any participant suffering harm as a result of their involvement in the research (Newline Underwriting Management Ltd, at Lloyd’s of London). In the UK, NHS indemnity operates in respect of the clinical treatment that is provided.

6.4 Local Clinical Centres The study will be conducted at multiple hospitals (LCCs) within each region. At each LCC, a lead investigator will be responsible for trial activities but much of the work will be carried out by medical staff attending patients with COVID-19 within the hospital and by hospital research nurses, medical students and other staff with appropriate education, training, and experience. Where LCCs plan to recruit children the principal investigator will co-opt support from a local paediatrician and/or neonatologists to oversee the management of children and infants in the trial.

6.5 Supply of study treatments For licensed treatments (e.g. lopinavir-ritonavir, corticosteroids, tocilizumab) all aspects of treatment supply, storage, and management will be in accordance with standard local policy and practice for prescription medications. Treatment issue to randomised participants will be by prescription. Such study treatments will not be labelled other than as required for routine clinical use. They will be stored alongside other routine medications with no additional monitoring. No accountability records will be kept beyond those used for routine prescriptions.

For unlicensed treatments, manufacture, packaging, labelling and delivery will be the responsibility of the pharmaceutical company and, in the UK, the Department of Health and Social Care. Each LCC will maintain an accountability log and will be responsible for the storage and issue of study treatment. If treatments require storage at a specific temperature, LCCs can use existing temperature-controlled facilities and associated monitoring. Treatment issue to randomised participants will be in accordance with local practice (and may be in line with the processes required for routine prescriptions or compassionate use).

For convalescent plasma in the UK, manufacture, packaging, and delivery will be the responsibility of the relevant UK Blood Service (NHS Blood and Transplant for England, Welsh Blood Service for Wales, Scottish National Blood Transfusion Service for Scotland, and the Northern Ireland Blood Transfusion Service for Northern Ireland). Convalescent plasma will be labelled in accordance with regulatory requirements and the unit will be issued to the ward for a named patient in a bag marked for clinical trial use only.

Treatment will be issued to randomised participants by prescription.

Page 20 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

6.6 End of trial The end of the scheduled treatment phase is defined as the date of the last follow-up visit of the last participant. In the UK, it is intended to extend follow-up for a year or more beyond the final study visit through linkage to routine medical records and central medical databases. The end of the study is the date of the final data extraction from NHS Digital (anticipated to be 10 years after the last patient is enrolled).

6.7 Publications and reports The Trial Steering Committee will be responsible for drafting the main reports from the study and for review of any other reports. In general, papers initiated by the Trial Steering Committee (including the primary manuscript) will be written in the name of the RECOVERY Collaborative Group, with individual investigators named personally at the end of the report (or, to comply with journal requirements, in web-based material posted with the report).

The Trial Steering Committee will also establish a process by which proposals for additional publications (including from independent external researchers) are considered by the Trial Steering Committee. The Trial Steering Committee will facilitate the use of the study data and approval will not be unreasonably withheld. However, the Trial Steering Committee will need to be satisfied that any proposed publication is of high quality, honours the commitments made to the study participants in the consent documentation and ethical approvals, and is compliant with relevant legal and regulatory requirements (e.g. relating to data protection and privacy). The Trial Steering Committee will have the right to review and comment on any draft manuscripts prior to publication.

6.8 Substudies Proposals for substudies must be approved by the Trial Steering Committee and by the relevant ethics committee and competent authorities (where required) as a substantial amendment or separate study before they begin. In considering such proposals, the Trial Steering Committee will need to be satisfied that the proposed substudy is worthwhile and will not compromise the main study in any way (e.g. by impairing recruitment or the ability of the participating hospitals to provide care to all patients under their care).

Page 21 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

7 VERSION HISTORY

Version number Date Brief Description of Changes 1.0 13-Mar-2020 Initial version 2.0 21-Mar-2020 Addition of hydroxychloroquine. Administrative changes and other clarifications. 3.0 07-Apr-2020 Extension of eligibility to those with suspected COVID-19 Addition of arm. Addition of inclusion of adults who lack permanently lack capacity. Change to primary outcome from in-hospital death to death within 28 days of randomisation. 4.0 14-Apr-2020 Addition of second randomisation to tocilizumab vs. standard of care among patients with progressive COVID-19. 5.0 24-Apr-2020 Addition of children to study population. 6.0 14-May-2020 Addition of convalescent plasma 7.0 18-Jun-2020 Allowance of randomisation in part B of main randomisation without part A. Removal of hydroxychloroquine and dexamethasone treatment arms. 8.0 03-Jul-2020 Removal of lopinavir-ritonavir Addition of intravenous immunoglobulin arm for children Changes to corticosteroid dosing for children. Addition of baseline serum sample in convalescent plasma randomisation 9.0 10-Sep-2020 Addition of synthetic neutralizing antibodies Additional baseline data collection Addition of countries outside UK 9.1 18-Sep-2020 Addition of information about vaccination of children of pregnant mothers receiving REGN10933+REGN10987 9.2 [not submitted in 15-Oct-2020 Additional information for countries outside UK UK] 10.0 26-Oct-2020 Addition of main randomisation part C General updates to avoid duplication and improve clarity 10.1 01-Nov-2020 Additional information for pregnant women 11.0 19-Nov-2020 Addition of colchicine to main randomisation part A Removal of azithromycin from main randomization part A Change in randomisation ratio in main randomisation part A from 2:1 to 1:1 11.1 21-Nov-2020 Clarification of colchicine age thresholds 11.2 [not submitted in 01-Dec-2020 Addition of modified aspirin dose if 150mg not available UK] 12.0 10-Dec-2020 Allow second randomisation of children without first randomisation 12.1 16-Dec-2020 Clarification of change in V12.0

Page 22 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

8 APPENDICES

8.1 Appendix 1: Information about the treatment arms

All patients will receive usual care in the participating hospital.

[UK only] Corticosteroids: Favourable modulation of the immune response is considered one of the possible mechanisms by which corticosteroids might be beneficial in the treatment of severe acute respiratory coronavirus infections, including COVID-19, SARS and MERS. Common to severe cases of these infections is the presence of hypercytokinemia (a cytokine ‘storm’) and development of acute lung injury or acute respiratory distress syndrome (ARDS).13-16 Pathologically, diffuse alveolar damage is found in patients who die from these infections.17 A growing volume of clinical trial data from patients with severe community acquired pneumonia, ARDS and septic shock suggest benefit from low-to-moderate dose corticosteroids in relation to mortality and length of stay.18-20

In trials of low-to-moderate doses of corticosteroids, the main adverse effect has been hyperglycaemia.19,21 A systematic review of (mainly low-dose) corticosteroid trials in severe sepsis and septic shock did not identify any increased risk of gastroduodenal bleeding, superinfection or neuromuscular weakness; an association with an increased risk of hyperglycaemia (RR 1.16, 95% CI 1.07 to 1.25) and hypernatraemia (RR 1.61, 95% CI 1.26 to 2.06) was noted.22

Methylprednisolone is a corticosteroid with mainly glucocorticoid activity. It is used in the treatment of conditions in which rapid and intense corticosteroid effect is required. Its licensed indications for paediatrics include a wide range of conditions including inflammatory disorders, allergic disorders, draft rejection reactions, severe erythema multiforme, juvenile idiopathic arthritis, and many others. In the paediatric population, a dosage of 10 mg/kg/day to a maximum of 1 g/day for up to 3 days is recommended in the treatment of graft rejection reactions following transplantation. A higher dosage of 30 mg/kg/day to a maximum of 1 g/day for up to 3 days is recommended for the treatment of haematological, rheumatic, renal and dermatological conditions (Source: British National Formulary for Children). Storage should be as per conditions in the Summary of Product Characteristics.

PIMS-TS is associated with a hyper-inflammatory state with elevated ESR, C-reactive protein, D-dimers, lactate dehydrogenase, ferritin, and increased levels of pro-inflammatory cytokines including as IL-1 and IL-6. While there is a pharmacological basis for using high dose methylprednisolone, the Delphi consensus process conducted by NHS England identified equipoise for its use in the treatment of PIMS-TS.

Colchicine: Colchicine inhibits cellular transport and mitosis by binding to tubulin and preventing its polymerisation as part of the cytoskeleton transport system. As a consequence, colchicine has a wide range of anti-inflammatory effects, including inhibition of certain inflammasomes (cytosolic pattern recognition receptor systems that are activated in response to detection of pathogens in the cytosol).23,24 There is evidence that inflammasomes are activated in COVID-19, and the degree of activation is correlated with disease severity.25 Colchicine has been widely used for treatment of gout and pericarditis, and there is evidence of cardiovascular benefit in patients with coronary artery disease. The

Page 23 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

UK COVID-19 Therapeutics Advisory Panel has recommended that RECOVERY assess colchicine.

[UK only] Intravenous immunoglobulin (IVIg): IVIg is human normal immunoglobulin, available in a number of different preparations in routine NHS practice. The NHS England consensus process has established intravenous immunoglobulin as the interim first line treatment in non-shocked COVID-associated PIMS-TS and also that there is need for evaluation of intravenous immunoglobulin and corticosteroid in the initial management of PIMS-TS. In the similar but different disease process known as Kawasaki Diseases, randomised controlled trials and meta-analyses have demonstrated that early recognition and treatment of KD with IVIg (and aspirin) reduces the occurrence of coronary artery aneurysms. Current published guidelines recommend a dose of 2 g/kg IVIg given as a single infusion, as this has been shown to reduce the coronary artery aneurysm rate compared to a lower divided dose regimen.26

IVIg is licensed for immunomodulation in adults, children and adolescents (0-18 years) in a number of clinical conditions including but not limited to primary immune thrombocytopenia, Guillain Barré syndrome, Kawasaki disease (in association with aspirin), chronic inflammatory demyelinating polyradiculoneuropathy and multifocal motor neuropathy.

Tocilizumab is a monoclonal antibody that binds to the receptor for IL-6, blocking IL-6 signalling and reduces inflammation. Tocilizumab is licensed for use in patients with rheumatoid arthritis and for use in people aged at least 2 years with chimeric antigen receptor (CAR) T cell-induced severe or life-threatening cytokine release syndrome.

Severe COVID-19 is associated with a hyper-inflammatory state with elevated ESR, C- reactive protein, D-dimers, lactate dehydrogenase, ferritin, and increased levels of pro- inflammatory cytokines including as IL-1 and IL-6.4,9,27 There have been published and unpublished (pre-print) case series reports of the successful treatment of COVID-19 patients with IL-6 inhibitors.27,28 IL-6 inhibitors have not been evaluated for the treatment of COVID- 19 in randomised controlled trials.

[UK only] Convalescent plasma: Convalescent plasma treatment, containing high titres of polyclonal antibody, has been used to treat severe viral pneumonias. Many studies have been small or poorly controlled but have reported beneficial effects in avian influenza29-31, influenza A (H1N1) infections in 1915-191732 and 2009/201033,34, and seasonal influenza B35. More relevant to SARS-CoV-2, a systematic review of convalescent plasma treatment in SARS-CoV infections in 2003 identified eight observational studies that all reported improved mortality associated with the use of convalescent plasma – infected patients received various amounts of convalescent plasma.36 Recent studies in seasonal influenza A and in MERS-CoV highlight the importance of high avidity and high titre antibodies respectively.37,38

Convalescent plasma therapy had been given to at least 245 COVID-19 patients by the end of February 2020, and, according to a Chinese health official, 91 cases had shown improvement in clinical indicators and symptoms. Five small case series (26 patients in total) have been published that report the use of convalescent plasma in people with COVID-19 infection.39-43 These studies have reported clinical and radiological improvements after treatment with convalescent plasma. However, these small uncontrolled studies have Page 24 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

significant flaws and the reported effects are unreliable. Convalescent plasma is currently being tested in the REMAP-CAP trial among patients on intensive care units.

[UK only] Synthetic neutralising antibodies (REGN-COV2): Synthetic monoclonal antibodies (mAbs) have been demonstrated to be safe and effective in viral disease when used as prophylaxis (respiratory syncytial virus and rabies) and treatment (Ebola virus disease).44,45 Anti-SARS-CoV-2 mAbs are designed to bind to and neutralise the virus. In addition, mAbs may have additional effector functions (antibody dependent phagocytosis and cytotoxicity) through binding to SARS-CoV-2 spike protein expressed on the surface of cells. Anti-SARS-CoV-2 spike protein neutralizing mAbs have demonstrated in vivo efficacy in both therapeutic and prophylactic settings in mouse, and non-human primates models, with decreases in viral load and lung pathology.2,46,47

Regeneron has developed 2 non-competing, high-affinity human IgG1 anti-SARS-CoV-2 mAbs, REGN10933 and REGN10987 that bind specifically to the receptor binding domain of the spike glycoprotein of SARS-CoV-2, blocking viral entry into host cells.48,49 REGN10933 and REGN10987 are both potent neutralizing antibodies that block the interaction between the spike protein and its canonical receptor angiotensin-converting enzyme 2. REGN10933 and REGN10987 are intended to be utilized as a combination treatment, known as REGN-COV2, and should not be used individually as monotherapy. A combination of antibodies that bind to non-overlapping epitopes may minimize the likelihood of loss of antiviral activity due to naturally circulating viral variants or development of escape mutants under drug pressure. In animal studies (rhesus macaques and hamsters) the antibody cocktail (REGN10933+REGN10987) reduced virus load in lower and upper airway and decreased virus induced pathological sequelae when administered prophylactically or therapeutically.50

Aspirin: Patients with COVID-19 appear to be at high risk of thromboembolism.51 Classical risk factors for thromboembolism are common in the COVID-19 hospitalised population, but the relatively low incidence of deep vein thrombosis compared to the incidence of pulmonary embolism (and the often peripheral location of the pulmonary emboli observed) suggests that inflammation and associated endothelial injury and platelet activation may be an important cause of thromboembolism in this patient population.51,52 Therefore antiplatelet therapy is a potential thromboprophylactic therapy in COVID-19. It is also being tested in the REMAP-CAP trial.

Page 25 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

8.2 Appendix 2: Drug specific contraindications and cautions

Corticosteroid (children only)  Known contra-indication to short-term corticosteroid.

Colchicine (men ≥18 years old and women ≥55 years old only) Contraindications:  Female participants <55 years old (as contraindicated in women of child-bearing potential)  Severe hepatic impairment (defined as requiring ongoing specialist care)  Significant cytopaenia (e.g. neutrophil count <1.0 x109/L; platelet count <50 x109/L; reticulocyte count <20 x109/L [if available])  Concomitant use of strong CYP3A4 inhibitor (e.g. clarithromycin, erythromycin, systemic azole antifungal, HIV protease inhibitor) or P-gp inhibitor (e.g. ciclosporin, verapamil, quinidine).  Hypersensitivity to lactose

Cautions: dose frequency should be halved (i.e. 1 mg at randomisation, 500 mcg 12 hours later then 500 mcg once daily) in the following circumstances.  Concomitant use of moderate CYP3A4 inhibitor (e.g. diltiazem)  Renal impairment: eGFR <30 mL/min/1.73m2 (either chronic or acute)  Estimated body weight <70 kg (If >1 of these is present, investigator should consider not including colchicine in randomisation.)

Participants allocated colchicine should have full blood counts monitored at a frequency determined by their clinician.

Intravenous Immunoglobulin (children only)  Hypersensitivity to the active substance (human immunoglobulins) or to any of the excipients  Patients with selective IgA deficiency who developed antibodies to IgA, as administering an IgA-containing product can result in anaphylaxis  Hyperprolinaemia type I or II. Potential complications can often be avoided by ensuring that participants: i. are carefully monitored for any symptoms throughout the infusion period; ii. have urine output and serum creatinine levels monitored; and iii. avoid concomitant use of loop diuretics. Such monitoring should occur regularly during the admission, at a frequency appropriate to the illness of the child.

Aspirin  Age <18 years old  Known hypersensitivity to aspirin  Recent major bleeding that precludes use of aspirin in opinion of managing physician Page 26 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

 Current use of aspirin, clopidogrel or other antiplatelet therapy

Tocilizumab  Known hypersensitivity to tocilizumab.  Evidence of active TB infectioni  Clear evidence of active bacterial, fungal, viral, or other infection (besides COVID- 19) (Note: Pregnancy and breastfeeding are not exclusion criteria.)

Convalescent plasma  Known moderate or severe allergy to blood components*  Not willing to receive a blood product* Synthetic neutralising antibodies (REGN-COV2)  Intravenous immunoglobulin treatment during current admission*  Age <12 years old or child with weight <40kg* (Note: Pregnancy and breastfeeding are not exclusion criteria.)

The infusion of synthetic neutralising antibodies should be interrupted if any of the following are observed (or worsen during the infusion): sustained/severe cough, rigors/chills, rash, pruritus, urticaria, diaphoresis, hypotension, dyspnoea, vomiting, or flushing. The reactions should be treated symptomatically, and the infusion may be restarted at 50% of the original rate once all symptoms have ceased (or returned to baseline) and at the discretion of the managing physician. If the managing physician feels there is medical need for treatment or discontinuation of the infusion other than described above, they should use clinical judgement to provide appropriate response according to typical clinical practice.

Pregnant women that are administered REGN10933 and REGN10987 must be advised that live vaccines should be avoided in children with in utero exposure to biologics for at least the first 6 months of life.

* If these conditions are recorded on the baseline case report form, patients will be ineligible for randomisation to that arm of the study.

Note: This study is being conducted within hospitals. Therefore use of medication will be subject to standard medication reviews (typically within 48 hours of enrolment) and clinical assessments (including appropriate blood tests) which will guide modifications to both the study treatment and use of concomitant medication (e.g. in the case of potential drug interactions). The doctor may decide whether it is appropriate to stop such medications temporarily to allow the patient to complete the course of their assigned intervention.

Although all available data on use in pregnancy are reassuring, since the effect of some of the treatments on unborn babies is uncertain, female participants who are not already pregnant will be advised that they should not get pregnant within 3 months of the completion of trial treatment(s).

i Note: The risk of reactivation of latent tuberculosis with tocilizumab is considered to be extremely small.

Page 27 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Page 28 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

8.3 Appendix 3: Paediatric dosing information

Children (aged <18 years old) will be recruited in the UK only.

Main Randomisation Part A Arm Route Weight # Dose (Duration for all arms = 10 days or until discharge from hospital) - - - No additional treatment

Corticosteroid Intravenous All Neonates/infants with a corrected gestational - Solution for injection* Including age of ≤44 weeks - Powder for solution for pre-term injection* neonates Hydrocortisone (IV): - 0.5 mg/kg every 12 hours for 7 days and then *various strengths available 0.5mg/kg once daily for 3 days

Intravenous All For all other children (with PIMS-TS):

Methylprednisolone sodium succinate 10 mg/kg (as base) once daily for 3 days (max 1 gram)

No additional oral corticosteroid should be prescribed to follow the 3 day treatment course.

Human normal Intravenous All For children with corrected gestational age >44 immunoglobulin (IVIg) weeks and <18 years with PIMS-TS phenotype:

- solution for infusion 2 g/kg as a single dose. (Dose should be based on ideal body weight in line with NHS England *various strengths available guidance.)

# Weight to be rounded to the nearest kg unless dosage expressed as mg/kg or mL/kg.

Page 29 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Main Randomisation Part B Convalescent Plasma Intravenous 5 mL/kg of ABO compatible convalescent plasma intravenous up to standard adult dose of 275 mLs per day on study days 1 and 2.

Minimum of 12 hour interval between 1st and 2nd units.

Convalescent plasma for neonates and infants up to one year of age needs to be ordered on a named patient basis from the relevant National Blood Service to ensure the unit meets neonatal requirements. Data transfer storage and retention will be in line with NHSBT standard procedures and protocols. Synthetic neutralising Intravenous ≥12 years 8 g (4 g of each monoclonal antibody) antibodies And (REGN10933 + REGN10987) ≥40 kg

Second stage randomisation (Patients < 1 year of age will NOT be eligible)

Arm Route Weight Dose

No additional treatment - - - Tocilizumab Intravenous Infants < 1 year excluded

< 30 kg 12 mg/kg A second dose may be given ≥12 and 24 hours later if, in the opinion of the attending clinicians, the patient’s condition has not improved.

≥ 30 kg 8 mg/kg (max 800 mg) A second dose may be given ≥12 and 24 hours later if, in the opinion of the attending clinicians, the patient’s condition has not improved.

Page 30 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

8.4 Appendix 4: Use of IMPs in pregnant and breastfeeding women All trial drugs (except REGN-COV2) have been used in pregnant women with pre-existing medical disorders where benefits outweigh the risks to fetus or woman, including in the first trimester. The existing data related to each drug is summarized below.

Colchicine Colchicine is contraindicated in pregnant or breastfeeding women.

Convalescent plasma Convalescent plasma is plasma from people who had confirmed COVID-19 (SARS-Cov-2) infection, and have now recovered and been free of the infection for 28 days. The plasma contains antibodies that their immune systems have produced in fighting the virus. It is hoped that giving this plasma will help speed up recovery of a patient with active infection and improve their chances of survival. Plasma is already used as a treatment in pregnant patients who are bleeding,53 or have particular blood conditions.54,55 The plasma being used in this trial is from a selected donor and hopefully contains anti-SARS-Cov-2 antibodies, but is otherwise no different. Plasma infusions can occasionally cause side effects. Mostly this is a rise in temperature, itching or a rash, and in very extreme cases, anaphylaxis. Other potential complications include breathlessness and changes in blood pressure. Monitoring of pulse and blood pressure takes place before and after the infusion. There is no risk of miscarriage or fetal loss, preterm birth, preterm rupture of membranes, perinatal mortality or low birthweight, from plasma transfusions and there are no concerns with breast feeding.

REGN-COV2 Monoclonal antibodies Monoclonal antibodies have been used as therapeutic agents in pregnancy over recent years, for a variety of conditions. Human monoclonal antibodies in use in pregnancy include anti-TNF agents, such as adalimumab, indicated for a variety of chronic inflammatory diseases such as rheumatoid arthritis and inflammatory bowel disease. Data have recently accumulated from a variety of cohort and registry studies indicating that such exposure in pregnancy was not associated with an increased risk for adverse pregnancy outcomes, when compared to unexposed pregnancies with the same underlying medical diseases.56 This is supported by a consensus report on immunosuppressives and biologics during pregnancy and lactation, confirming no evidence of elevated adverse pregnancy outcomes or malformation risks.57 Some monoclonal antibodies are transported across the placenta (and may also enter breast milk) but as REGN10933 and REGN10987 do not have any human targets such exposure should not be associated with risk of harm. Pregnant women, just like other patients with COVID-19, are at significant risk from the infection itself (particularly those in the third trimester.58,59 All pregnant women in RECOVERY are entered into the UK Obstetric Surveillance System which follows all pregnancies to their conclusion.59 Given the early safety experience with REGN10933+REGN10987 it would appear appropriate not to exclude pregnant women from this aspect of the trial (as such exclusion would inhibit the development of treatments for this population).60

Aspirin Aspirin is widely used for the prevention of pre-eclampsia in pregnant women at increased risk of the disease. A recent Cochrane meta-analysis on this topic included seventy-seven trials (40,249 women) taking aspirin at doses between 60 and 150mg daily.61 In most trials, aspirin was started from 12 weeks’ gestation, although a more recent meta-analysis has reported eight trials (1426 women) in which aspirin was initiated in the first trimester.62 In

Page 31 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

light of the clear evidence of effectiveness, 75-150mg aspirin is recommended for pre- eclampsia prophylaxis in NICE guidelines for management of hypertension in pregnancy (NG133), and in the NHS England document ‘Saving Babies’ Lives for women at increased risk of placental dysfunction disorders.63,64 There is some ongoing uncertainty as to the optimal dose (75mg vs. 150mg) for pre-eclampsia prophylaxis, but both doses are in widespread clinical use in pregnancy in the UK for these indications and in other conditions (e.g. in pregnant women with antiphospholipid syndrome).

Tocilizumab Two pharmaceutical global safety registry database studies have reported on tocilizumab use in pregnancy, including outcomes from 288 pregnancies 65 and 61 pregnancies,66 typically for rheumatoid or other arthritides, and with the majority having received the drug in the first trimester. These data suggest that the rates of congenital abnormality, spontaneous pregnancy loss and other adverse outcomes were not higher than in the general population.66 Small studies have shown that tocilizumab is transferred to the fetus with serum concentrations approximately 7-fold lower than those observed in maternal serum at the time of birth.67 Very low concentrations of tocilizumab are identified in breast milk and no drug is transferred into the serum of breast fed infants.67,68 Women should be advised that if treated after 20 weeks’ gestation, their infant should not be immunised with live vaccines (rotavirus and BCG) for the first 6 months of life. All non-live vaccinations are safe and should be undertaken.69

Page 32 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

8.5 Appendix 5: Organisational Structure and Responsibilities

Chief Investigator The Chief Investigator has overall responsibility for:

(i) Design and conduct of the Study in collaboration with the Trial Steering Committee; (ii) Preparation of the Protocol and subsequent revisions;

Trial Steering Committee The Trial Steering Committee (see Section 8.6 for list of members) is responsible for:

(i) Agreement of the final Protocol and the Data Analysis Plans; (ii) Reviewing progress of the study and, if necessary, deciding on Protocol changes; (iii) Review and approval of study publications and substudy proposals; (iv) Reviewing new studies that may be of relevance.

Regional (South East Asia) Steering Committee The regional SEA Steering Committee (see Section 8.6 for list of members) is responsible for:

(i) Reviewing progress of the study in South East Asia; (ii) Review of study publications and substudy proposals; (iii) Considering potential new therapies to be included in South East Asia; (iv) Assisting RCC in selection of LCCs (v) Reviewing new studies that may be of relevance.

Data Monitoring Committee The independent Data Monitoring Committee is responsible for:

(i) Reviewing unblinded interim analyses according to the Protocol; (ii) Advising the Steering Committee if, in their view, the randomised data provide evidence that may warrant a change in the protocol (e.g. modification or cessation of one or more of the treatment arms).

Central Coordinating Office (CCO) The CCO is responsible for the overall coordination of the Study, including:

(i) Study planning and organisation of Steering Committee meetings; (ii) Ensuring necessary regulatory and ethics committee approvals; (iii) Development of Standard Operating Procedures and computer systems (iv) Monitoring overall progress of the study; (v) Provision of study materials to RCCs/LCCs; (vi) Monitoring and reporting safety information in line with the protocol and regulatory requirements; (vii) Dealing with technical, medical and administrative queries from LCCs.

Page 33 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

Regional Coordinating Centre (RCC) The RCCs are responsible for:

(i) Ensuring necessary regulatory and ethics committee approvals; (ii) Provision of study materials to LCCs; (iii) Dealing with technical, medical and administrative queries from LCCs.

Local Clinical Centres (LCC) The LCC lead investigator and LCC clinic staff are responsible for:

(i) Obtaining all relevant local permissions (assisted by the CCO) (ii) All trial activities at the LCC, including appropriate training and supervision for clinical staff (iii) Conducting trial procedures at the LCC in line with all relevant local policies and procedures; (iv) Dealing with enquiries from participants and others.

Page 34 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

8.6 Appendix 5: Organisational Details

STEERING COMMITTEE (Major organisational and policy decisions, and scientific advice; blinded to treatment allocation)

Chief Investigator Peter Horby Deputy Chief Investigator Martin Landray Clinical Trial Unit Lead Richard Haynes Co-investigators Kenneth Baillie (Scotland Lead), Maya Buch, Lucy Chappell, Saul Faust, Thomas Jaki, Katie Jeffery, Edmund Juszczak, Wei Shen Lim, Marion Mafham, Alan Montgomery, Andrew Mumford, Kathy Rowan, Guy Thwaites, Jeremy Day (South East Asia Leads)

South East Asia Steering Committee (Members TBD)

Regional Lead Investigators Guy Thwaites, Jeremy Day Country Lead Investigators: TBD (Nepal), TBD (), TBD () MOH or local country representatives: TBD (Nepal), TBD (VietNam), TBD (Indonesia) Independent members: TBD (3 members)

DATA MONITORING COMMITTEE (Interim analyses and response to specific concerns)

Chair Peter Sandercock Members Janet Darbyshire, David DeMets, Robert Fowler, David Lalloo, Ian Roberts, Janet Wittes Statisticians (non-voting) Jonathan Emberson, Natalie Staplin

Page 35 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

9 REFERENCES 1. Zhu N, Zhang D, Wang W, et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N Engl J Med 2020;382:727-33. 2. Shi R, Shan C, Duan X, et al. A human neutralizing antibody targets the receptor-binding site of SARS- CoV-2. Nature 2020;584:120-4. 3. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497-506. 4. Wang D, Hu B, Hu C, et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA 2020. 5. Whittaker E, Bamford A, Kenny J, et al. Clinical Characteristics of 58 Children With a Pediatric Inflammatory Multisystem Syndrome Temporally Associated With SARS-CoV-2. JAMA 2020. 6. Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020;395:507-13. 7. Mehta P, McAuley DF, Brown M, et al. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 2020;395:1033-4. 8. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med 2020;46:846-8. 9. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID- 19 in Wuhan, China: a retrospective cohort study. Lancet 2020;395:1054-62. 10. Venet D, Doffagne E, Burzykowski T, et al. A statistical approach to central monitoring of data quality in clinical trials. Clin Trials 2012;9:705-13. 11. Oversight of Clinical Investigations--A Risk-Based Approach to Monitoring. 2013. (Accessed 18 August 2017, at https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM269919.p df.) 12. Administration. USDoHaHSFaD. Guidance for Industry Part 11, Electronic Records; Electronic Signatures — Scope and Application. 2003. 13. Lau SKP, Lau CCY, Chan KH, et al. Delayed induction of proinflammatory cytokines and suppression of innate antiviral response by the novel Middle East respiratory syndrome coronavirus: implications for pathogenesis and treatment. J Gen Virol 2013;94:2679-90. 14. de Jong MD, Simmons CP, Thanh TT, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med 2006;12:1203-7. 15. Liu Q, Zhou YH, Yang ZQ. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol 2016;13:3-10. 16. Short KR, Veeris R, Leijten LM, et al. Proinflammatory Cytokine Responses in Extra-Respiratory Tissues During Severe Influenza. J Infect Dis 2017;216:829-33. 17. Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 2020;8:420-2. 18. Rochwerg B, Oczkowski SJ, Siemieniuk RAC, et al. Corticosteroids in Sepsis: An Updated Systematic Review and Meta-Analysis. Crit Care Med 2018;46:1411-20. 19. Villar J, Ferrando C, Martinez D, et al. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med 2020;8:267-76. 20. Siemieniuk RA, Meade MO, Alonso-Coello P, et al. Corticosteroid Therapy for Patients Hospitalized With Community-Acquired Pneumonia: A Systematic Review and Meta-analysis. Ann Intern Med 2015;163:519-28. 21. Meijvis SC, Hardeman H, Remmelts HH, et al. Dexamethasone and length of hospital stay in patients with community-acquired pneumonia: a randomised, double-blind, placebo-controlled trial. Lancet 2011;377:2023-30. 22. Annane D, Bellissant E, Bollaert PE, et al. Corticosteroids in the treatment of severe sepsis and septic shock in adults: a systematic review. JAMA 2009;301:2362-75. 23. Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol 2016;16:407-20. 24. Dalbeth N, Lauterio TJ, Wolfe HR. Mechanism of action of colchicine in the treatment of gout. Clin Ther 2014;36:1465-79. 25. Rodrigues TS, Keyla SG, Ishimoto AY, et al. Inflammasome activation in COVID-19 patients. medRxiv 2020. 26. Eleftheriou D, Levin M, Shingadia D, Tulloh R, Klein NJ, Brogan PA. Management of Kawasaki disease. Arch Dis Child 2014;99:74-83.

Page 36 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

27. Zhang W, Zhao Y, Zhang F, et al. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The Perspectives of clinical immunologists from China. Clin Immunol 2020;214:108393. 28. Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. The cytokine release syndrome (CRS) of severe COVID- 19 and Interleukin-6 receptor (IL-6R) antagonist Tocilizumab may be the key to reduce the mortality. Int J Antimicrob Agents 2020:105954. 29. Zhou B, Zhong N, Guan Y. Treatment with convalescent plasma for influenza A (H5N1) infection. N Engl J Med 2007;357:1450-1. 30. Wu XX, Gao HN, Wu HB, Peng XM, Ou HL, Li LJ. Successful treatment of avian-origin influenza A (H7N9) infection using convalescent plasma. Int J Infect Dis 2015;41:3-5. 31. Kong LK, Zhou BP. Successful treatment of avian influenza with convalescent plasma. Hong Kong Med J 2006;12:489. 32. Luke TC, Kilbane EM, Jackson JL, Hoffman SL. Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann Intern Med 2006;145:599-609. 33. Hung IF, To KK, Lee CK, et al. Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza A (H1N1) 2009 virus infection. Clin Infect Dis 2011;52:447-56. 34. Hung IFN, To KKW, Lee CK, et al. Hyperimmune IV immunoglobulin treatment: a multicenter double- blind randomized controlled trial for patients with severe 2009 influenza A(H1N1) infection. Chest 2013;144:464-73. 35. Davey RT, Jr., Fernandez-Cruz E, Markowitz N, et al. Anti-influenza hyperimmune intravenous immunoglobulin for adults with influenza A or B infection (FLU-IVIG): a double-blind, randomised, placebo- controlled trial. Lancet Respir Med 2019;7:951-63. 36. Mair-Jenkins J, Saavedra-Campos M, Baillie JK, et al. The effectiveness of convalescent plasma and hyperimmune immunoglobulin for the treatment of severe acute respiratory infections of viral etiology: a systematic review and exploratory meta-analysis. J Infect Dis 2015;211:80-90. 37. Beigel JH, Aga E, Elie-Turenne MC, et al. Anti-influenza immune plasma for the treatment of patients with severe influenza A: a randomised, double-blind, phase 3 trial. Lancet Respir Med 2019;7:941-50. 38. Arabi YM, Hajeer AH, Luke T, et al. Feasibility of Using Convalescent Plasma Immunotherapy for MERS-CoV Infection, Saudi Arabia. Emerg Infect Dis 2016;22:1554-61. 39. Ahn JY, Sohn Y, Lee SH, et al. Use of Convalescent Plasma Therapy in Two COVID-19 Patients with Acute Respiratory Distress Syndrome in Korea. J Korean Med Sci 2020;35:e149. 40. Zhang B, Liu S, Tan T, et al. Treatment With Convalescent Plasma for Critically Ill Patients With SARS- CoV-2 Infection. Chest 2020. 41. Ye M, Fu D, Ren Y, et al. Treatment with convalescent plasma for COVID-19 patients in Wuhan, China. J Med Virol 2020. 42. Shen C, Wang Z, Zhao F, et al. Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma. JAMA 2020. 43. Duan K, Liu B, Li C, et al. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc Natl Acad Sci U S A 2020. 44. Laustsen AH. How can monoclonal antibodies be harnessed against neglected tropical diseases and other infectious diseases? Expert Opin Drug Discov 2019;14:1103-12. 45. Mulangu S, Dodd LE, Davey RT, et al. A Randomized, Controlled Trial of Ebola Virus Disease Therapeutics. New England Journal of Medicine 2019;381:2293-303. 46. Cao Y, Su B, Guo X, et al. Potent Neutralizing Antibodies against SARS-CoV-2 Identified by High- Throughput Single-Cell Sequencing of Convalescent Patients' B Cells. Cell 2020;182:73-84 e16. 47. Zost SJ, Gilchuk P, Case JB, et al. Potently neutralizing and protective human antibodies against SARS-CoV-2. Nature 2020. 48. Hansen J, Baum A, Pascal KE, et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science 2020. 49. Regeneron Pharmaceuticals Inc. Investigator's Brochure REGN10933+REGN10987. 2020. 50. Baum A, Copin R, Ajithdoss D, et al. REGN-COV2 antibody cocktail prevents and treats SARS-CoV- 2 infection in rhesus macaques and hamsters. bioRxiv 2020:2020.08.02.233320. 51. Desborough MJR, Doyle AJ, Griffiths A, Retter A, Breen KA, Hunt BJ. Image-proven thromboembolism in patients with severe COVID-19 in a tertiary critical care unit in the United Kingdom. Thromb Res 2020;193:1- 4. 52. Taus F, Salvagno G, Cane S, et al. Platelets Promote Thromboinflammation in SARS-CoV-2 Pneumonia. Arterioscler Thromb Vasc Biol 2020:ATVBAHA120315175. 53. Townsley DM. Hematologic complications of pregnancy. Semin Hematol 2013;50:222-31.

Page 37 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

54. Brooks JP, Radojicic C, Riedl MA, Newcomer SD, Banerji A, Hsu FI. Experience with Intravenous Plasma-Derived C1-Inhibitor in Pregnant Women with Hereditary Angioedema: A Systematic Literature Review. J Allergy Clin Immunol Pract 2020. 55. Scully M, Thomas M, Underwood M, et al. Thrombotic thrombocytopenic purpura and pregnancy: presentation, management, and subsequent pregnancy outcomes. Blood 2014;124:211-9. 56. Chambers CD, Johnson DL, Xu R, et al. Birth outcomes in women who have taken adalimumab in pregnancy: A prospective cohort study. PloS one 2019;14:e0223603. 57. Puchner A, Grochenig HP, Sautner J, et al. Immunosuppressives and biologics during pregnancy and lactation : A consensus report issued by the Austrian Societies of Gastroenterology and Hepatology and Rheumatology and Rehabilitation. Wien Klin Wochenschr 2019;131:29-44. 58. Allotey J, Stallings E, Bonet M, et al. Clinical manifestations, risk factors, and maternal and perinatal outcomes of coronavirus disease 2019 in pregnancy: living systematic review and meta-analysis. Bmj 2020;370:m3320. 59. Knight M, Bunch K, Vousden N, et al. Characteristics and outcomes of pregnant women admitted to hospital with confirmed SARS-CoV-2 infection in UK: national population based cohort study. Bmj 2020;369:m2107. 60. Knight M, Morris RK, Furniss J, Chappell LC. Include pregnant women in research-particularly covid- 19 research. Bmj 2020;370:m3305. 61. Duley L, Meher S, Hunter KE, Seidler AL, Askie LM. Antiplatelet agents for preventing pre-eclampsia and its complications. Cochrane Database Syst Rev 2019;2019. 62. Chaemsaithong P, Cuenca-Gomez D, Plana MN, Gil MM, Poon LC. Does low-dose aspirin initiated before 11 weeks' gestation reduce the rate of preeclampsia? Am J Obstet Gynecol 2020;222:437-50. 63. Webster K, Fishburn S, Maresh M, Findlay SC, Chappell LC, Guideline C. Diagnosis and management of hypertension in pregnancy: summary of updated NICE guidance. BMJ 2019;366:l5119. 64. IMPROVE-IT: Examining Outcomes in Subjects With Acute Coronary Syndrome: Vytorin (Ezetimibe/Simvastatin) vs Simvastatin. (Accessed 4 Aug 2013, 2013, at http://clinicaltrials.gov/ct2/show/NCT00202878.) 65. Hoeltzenbein M, Beck E, Rajwanshi R, et al. Tocilizumab use in pregnancy: Analysis of a global safety database including data from clinical trials and post-marketing data. Semin Arthritis Rheum 2016;46:238-45. 66. Nakajima K, Watanabe O, Mochizuki M, Nakasone A, Ishizuka N, Murashima A. Pregnancy outcomes after exposure to tocilizumab: A retrospective analysis of 61 patients in Japan. Mod Rheumatol 2016;26:667- 71. 67. Saito J, Yakuwa N, Kaneko K, et al. Tocilizumab during pregnancy and lactation: drug levels in maternal serum, cord blood, breast milk and infant serum. Rheumatology (Oxford) 2019;58:1505-7. 68. Saito J, Yakuwa N, Takai C, et al. Tocilizumab concentrations in maternal serum and breast milk during breastfeeding and a safety assessment in infants: a case study. Rheumatology (Oxford) 2018;57:1499- 501. 69. Flint J, Panchal S, Hurrell A, et al. BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding-Part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology (Oxford) 2016;55:1693-7.

Page 38 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21

10 CONTACT DETAILS

Website: www.recoverytrial.net (copies of this protocol and related forms and information can be downloaded)

RECOVERY Central Coordinating Office: Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF United Kingdom

Tel: +44 (0)800 1385451 E-mail: [email protected]

RECOVERY Vietnam: Oxford University Clinical Research Unit, Centre for Tropical Medicine, 764 Vo Van Kiet, District 5, Ho Chi Minh City, Vietnam Tel: +84 8 39241983 E-mail: [email protected]

RECOVERY Indonesia: Eijkman Oxford Clinical Research Unit (EOCRU), Eijkman Institute for Molecular Biology Jl. P. Diponegoro No. 69, Jakarta-Indonesia 10430 Tel: +62 21 31900971

RECOVERY Nepal: Clinical Trial Unit, Oxford University Clinical Research Unit-Nepal, Patan Academy of Health Sciences, Kathmandu, Nepal Tel : +977 01 5522295

To RANDOMISE a patient, visit:

Website: www.recoverytrial.net

Page 39 of 39 RECOVERY [V12.1 2020-12-16] ISRCTN50189673 EudraCT 2020-001113-21